Stchstc Gene Expn

download Stchstc Gene Expn

of 21

Transcript of Stchstc Gene Expn

  • 7/31/2019 Stchstc Gene Expn

    1/21

    Stochastic gene expression and its consequences

    Arjun Raj and Alexander van Oudenaarden*

    Department of Physics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA

    Abstract

    Gene expression is a fundamentally stochastic process, with randomness in transcription and

    translation leading to significant cell-to-cell variations in mRNA and protein levels. This variation

    appears in organisms ranging from microbes to metazoans and its characteristics depend both on

    the biophysical parameters governing gene expression and on gene network structure. Stochastic

    gene expression can have important consequences for cellular function, being beneficial in some

    contexts and harmful in others. These situations include stress response, pathogenesis,

    metabolism, development, cell cycle, circadian rhythms and aging.

    Introduction

    Life is a study in contrasts between randomness and determinism: from the chaos of

    biomolecular interactions to the precise coordination of development, living organisms are

    able to resolve these two seemingly contradictory aspects of their internal workings.

    Scientists often reconcile the stochastic and the deterministic by appealing to the statistics of

    large numbers, thus diminishing the importance of any one molecule in particular. However,

    cellular function often involves small numbers of molecules, of which perhaps the most

    important example is DNA. It is this molecule, usually present in just one or few copies per

    cell, that gives organisms their unique genetic identity. But what about genetically identical

    organisms grown in homogenous environments? To what degree are they unique?

    Increasingly, researchers have found that even genetically identical individuals can be very

    different, and that some of the most striking sources of this variability are random

    fluctuations in the expression of individual genes. Fundamentally, this is because the

    expression of a gene involves the discrete and inherently random biochemical reactions

    involved in the production of mRNAs and proteins. The fact that DNA (and hence the genes

    encoded therein) is present in very low numbers means that these fluctuations do not just

    average away but can instead lead to easily detectable differences between otherwise

    identical cells; in other words, gene expression must be thought of as a stochastic process.

    The experimental observation that the levels of gene expression vary from cell-to-cell is

    certainly not new. In 1957, Novick and Weiner showed that the production of beta-

    galactosidase in individual cells was highly variable and random, with induction increasing

    the proportion of cells expressing the enzyme rather than increasing every cell's expression

    level equally (Novick and Weiner, 1957). Such early studies were hindered, however, by thelack of reliable single-cell assays of gene expression. One of the first studies to use an

    expression reporter in single-cells to examine the stochastic underpinnings of expression

    variability is the pioneering work of Ko et al., 1990. They examined the effect of different

    doses of glucocorticoid on the expression of a glucocorticoid-responsive transgene encoding

    beta-galactosidase and found that the cell-to-cell variability in the expression of the

    transgene was surprising large. Moreover, increasing the dose led to an increased frequency

    *Corresponding author: [email protected]; 617-253-4446.

    NIH Public AccessAuthor ManuscriptCell. Author manuscript; available in PMC 2011 June 18.

    Published in final edited form as:

    Cell. 2008 October 17; 135(2): 216226. doi:10.1016/j.cell.2008.09.050.

    NIH-PAAu

    thorManuscript

    NIH-PAAuthorManuscript

    NIH-PAAuthorM

    anuscript

  • 7/31/2019 Stchstc Gene Expn

    2/21

    of cells displaying a high level of expression rather than a uniform increase in expression in

    every cell; that is, dose dependence was a consequence of changing the probability that an

    individual cell would express the gene at a high level.

    Yet despite the potential biological consequences of random cellular variability (Spudich

    and Koshland, 1976), several years would pass before theoretical work ignited much of the

    present interest in stochastic gene expression (McAdams and Arkin, 1997; Arkin et al. 1998)

    They modeled gene expression using a stochastic formulation of chemical kinetics derivedby Gillespie (1977), predicting that in some biologically realistic parameter ranges, protein

    numbers could fluctuate markedly within individual cells. Theythen extended their analysis

    to model the circuit underlying the decision between lysis and lysogeny of the phage-

    lambda, showing that stochastic effects in the expression of key regulators could explain

    why some cells activated the lytic pathway whereas others followed the lysogenic pathway.

    The notion that stochastic effects in gene expression could have important biological

    implications has motivated much research in the field and has only recently been explored

    experimentally.

    Since this early research, the study of stochastic gene expression has blossomed into a rich

    field, with researchers from a diverse set of backgrounds working on a wide range of

    problems. The field is also notable for its strong interplay between theory and experiment,

    with many scientists making significant contributions to both. In this review, we willdescribe these researchers' efforts to characterize the underlying phenomenon through a host

    of organisms using a variety of experimental and theoretical methods. We will then

    highlight some recent endeavors trying to tie stochastic gene expression to biological

    phenomena.

    Noisy bugs

    The first attempts to characterize stochastic gene expression were born from experiments in

    synthetic biology in which experimenters found that noisy behavior in gene expression was

    interfering with the operation of engineered genetic circuits. One example is the

    repressilator, a synthetic network of repressors that was capable of producing oscillations

    in gene expression (Elowitz and Leibler, 2000). The authors found that the oscillations were

    subject to marked fluctuations in their period and magnitude, and conjectured that stochasticeffects in gene expression were causing these effects. In another study explicitly aimed at

    controlling fluctuations, Becskei and Serrano (2000) showed that engineering a circuit with

    negative feedback could reduce cell-to-cell variability in expression. Although these

    experiments showed that noise in gene expression was important and could even be

    controlled, the molecular basis for the observed variability remained unclear.

    The first experiments to explore the causes of stochastic gene expression were the landmark

    studies of Elowitz et al. (2002) and Ozbudak et al. (2002). Elowitz et al. introduced the

    concepts of extrinsic and intrinsic noise in gene expression (analyzed mathematically by

    Swain et al., 2002). These ideas are perhaps most easily explained through example. In their

    experiments, Elowitz et al. quantified the variability in the expression from a promoter inE.

    coli by introducing two copies of the same promoter into the genome ofE. coli, one driving

    the expression of cyan fluorescent protein (CFP) and the other driving the expression ofyellow fluorescent protein (YFP) (Figures 1A and 1B). In this setup, extrinsic fluctuations

    are those that affect the expression of both copies of the gene equally in a given cell, such as

    variations in the numbers of RNA polymerases or ribosomes. Intrinsic fluctuations are those

    due to the randomness inherent to transcription and translation; being random, they should

    affect each copy of the gene independently, adding uncorrelated variations in levels of CFP

    and YFP levels (Figure 1C). They found that both sources of noise can be significant

    Raj and van Oudenaarden Page 2

    Cell. Author manuscript; available in PMC 2011 June 18.

    NIH-PAA

    uthorManuscript

    NIH-PAAuthorManuscript

    NIH-PAAuthor

    Manuscript

  • 7/31/2019 Stchstc Gene Expn

    3/21

    depending on the promoter. Later time-lapse measurements showed that in bacteria, the time

    scale for intrinsic fluctuations is less than 9 minutes, whereas extrinsic fluctuations exert

    their effects on time scales of about 40 minutes, or roughly the length of the cell cycle

    (Rosenfeld et al., 2005).

    Ozbudak et al. (2002) obsereved that variability in the expression of a gene expressing GFP

    driven by an inducible promoter inB. subtilis depended on the underlying biochemical rates

    of transcription and translation. In these experiments transcription rates were controlled byvarying the level of induction and the translation rate was altered by introducing mutations

    into the ribosomal binding site. This verified a stochastic theory of intrinsic noise they had

    developed predicting how noise in gene expression would change as these parameters were

    altered (Thattai and van Oudenaarden, 2001) (Figures 2A and 2B). In particular, the theory

    predicted that noise (measured by the standard deviation in protein expression level divided

    by the mean) would depend inversely on the rate of transcription but would not depend on

    the rate of translation. This is because proteins are produced in translational bursts from

    individual transcripts; the concept of bursts in gene expression continues to play an

    important role in current research, especially in higher eukaryotes.

    Recently, a set of exciting single-molecule experiments have observed translational bursts in

    individual living bacteria. To count the number of proteins per cell, Cai et al., 2006 used

    used two methods: one involving microfluidics, in which they quantified the number ofbeta-galactosidase enzymes in a cell by monitoring its enzymatic activity, and one involving

    direct visualization of single YFP molecules tethered to the cellular membrane (Yu et al.,

    2006). Both studies showed that proteins were synthesized in rapid, burst-like fashion.

    Another study (Golding et al., 2005) used the MS2-GFP method (Bertrand et al., 1998;

    Beach et al., 1999), which allows one to monitor the transcription of individual mRNA

    molecules in real time. This is accomplished by introducing a repeated sequence motif into

    the 3 untranslated region of the mRNA to which a fusion of the MS2 coat protein and GFP

    binds, thus rendering the mRNA molecule fluorescent. According to the model presented in

    Figure 3A, one would expect that mRNA molecules are produced at a steady rate according

    to the statistics of a Poisson process. The authors found, however, that the mRNA molecules

    were themselves produced in transcriptional bursts, as if the gene itself was randomly

    switching back and forth between transcriptionally active and inactive states. This findingmirrors results obtained for eukaryotes described below. It would be interesting to combine

    these different measurements of the dynamics of individual mRNAs and proteins, given the

    role that competition between translation and mRNA degradation may play in stochastic

    gene expression (Yarchuk et al., 1998).

    Eukaryotes and the burst hypothesis

    After these experiments in bacteria, researchers began to investigate stochastic gene

    expression in eukaryotes, initially focusing on yeast. Almost immediately, several reports

    seemed to indicate that the sources of variability in gene expression in yeast are different

    from those in bacteria in a number of important ways (Becskei et al., 2005; Blake et al.,

    2006; Blake et al., 2003; Raser and O'Shea, 2004). These studies all examined the

    relationship between the mean level of expression and the variation about that mean, arelationship that is in theory qualitatively different depending on the sources of noise. In all

    these studies, the relationship predicted by the simple model in Figure 2 was insufficient to

    explain the experimental observations. These observations were, however, compatible with

    models of transcriptional bursts in which the gene itself randomly transitioned between

    states of transcriptional activity and inactivity (Figure 3B). Such models of transcriptional

    bursting add another important source of stochasticity beyond random events in transcription

    Raj and van Oudenaarden Page 3

    Cell. Author manuscript; available in PMC 2011 June 18.

    NIH-PAA

    uthorManuscript

    NIH-PAAuthorManuscript

    NIH-PAAuthor

    Manuscript

  • 7/31/2019 Stchstc Gene Expn

    4/21

    and translation, which have now been analyzed theoretically in some detail (Friedman et al.,

    2006; Karmakar and Bose, 2004; Kepler and Elston, 2001; Pedraza and Paulsson, 2008).

    That such models are required to explain eukaryotic data but not most prokaryotic data (Cai

    et al., 2006; Maamar et al., 2007; Yu et al., 2006), with an important exception (Golding et

    al., 2005), strongly suggests that some regulator of gene expression specific to eukaryotes is

    responsible. The most likely candidate for this is remodeling of chromatin: when the

    surrounding chromatin is in an open, acetylated state, the gene is able to transcribe relativelyfreely, whereas when chromatin is in a condensed state, transcription is repressed. Although

    there is still no direct evidence that chromatin remodeling is responsible for stochastic

    changes in gene activity, several studies have tried to link chromatin-related events to

    stochastic gene expression by indirect means. These include positional effects like

    measuring correlations between proximally located genes (Becskei et al., 2005; Raj et al.,

    2006) or altering the behavior of chromatin remodeling agents (Raser and O'Shea, 2004; Xu

    et al., 2006). However, global studies of noise in yeast (Bar-Even et al., 2006; Newman et

    al., 2006) have shown that the presence of chromatin remodeling complexes is neither

    necessary nor sufficient for the expression of a gene to be noisy; also, factors such as the

    location and number of transcription factor binding sites can control noise (Murphy et al.,

    2007).

    In yeast noise in gene expression is primarily extrinsic in origin (Becskei et al., 2005;Colman-Lerner et al., 2005; Raser and O'Shea, 2004; Volfson et al., 2006), resulting in

    correlated fluctuations between different genes. Sources identified thus far for this extrinsic

    noise are cell size (Raser and O'Shea, 2004; Newman et al., 2006; Volfson et al., 2006),

    variations in common upstream factors (Becskei et al., 2005; Volfson et al., 2006) and

    chromosomal location (Becskei et al., 2005); by contrast, extrinsic variability in prokaryotic

    gene expression is thought to stem mostly from variations in upstream factors (Elowitz et

    al., 2002). There is some debate as to the role of differences in cell-cycle and cell size, with

    some data (Raser and O'Shea, 2004) showing that extrinsic variability remains even after

    controlling for these variables, whereas other data indicates that a stringent analysis of size

    and shape by flow cytometry can account for most of the extrinsic noise (Newman et al.,

    2006). Generally, one of the difficulties in studying extrinsic variability is its catchall nature:

    the lack of any specific mechanism makes its analysis rather phenomenological. Although

    there is some knowledge of the time scales over which extrinsic noise operates (Rosenfeld etal., 2005) and theoretical analyses of the effects that it might have (Shahrezaei et al., 2008)

    (Paulsson, 2004), understanding extrinsic noise remains an unresolved problem in the field.

    Higher eukaryotes: noisier than expected

    Meanwhile, work has begun on systematically examining cell-to-cell variability in gene

    expression in higher eukaryotes. A priori, one might expect that higher eukaryotes, with

    their larger size and numbers of molecules, might exhibit less variability than prokaryotes

    and yeast. On the other hand, the prevalence of transcriptionally-silenced heterochromatin

    would argue that slow, random events of gene activation and inactivation would lead to

    much larger fluctuations than in unicellular organisms. As it happens, the latter is the case,

    with a growing body of evidence that fluctuations in higher eukaryotes can be remarkably

    large.

    Interestingly, the study of expression variability in higher eukaryotes began well before the

    recent heightened interest in stochastic gene expression. Beginning with the aforementioned

    work of Ko et al. (1990), several other reports indicated that gene expression in mammalian

    cells was variable, stemming from short, rare events of active transcription (Ross et al.,

    1994; Newlands et al., 1998; Takasuka et al., 1998; White et al., 1995).

    Raj and van Oudenaarden Page 4

    Cell. Author manuscript; available in PMC 2011 June 18.

    NIH-PAA

    uthorManuscript

    NIH-PAAuthorManuscript

    NIH-PAAuthor

    Manuscript

  • 7/31/2019 Stchstc Gene Expn

    5/21

    Many of these early experiments were limited by the difficulties inherent to measuring gene

    expression in single-cells in higher eukaryotes. One problem is sensitivity: owing to their

    large cellular volumes, even moderately expressed fluorescent proteins can be difficult to

    detect. Another problem is the lack of tools available to manipulate these organisms

    genetically. To circumvent these problems, researchers have come up with many new ways

    of assaying gene expression at the single-cell level to measure cell-to-cell variability.

    One approach is to measure mRNAs rather than proteins. For instance, utilizing the MS2-GFP method of mRNA detection (Beach et al., 1999; Bertrand et al., 1998), Chubb et al.

    (2006) showed that a developmental gene inDictyostelium discoideum is transcribed in a

    pulsatile fashion, directly demonstrating the burst hypothesis by watching mRNAs

    accumulate and dissipate from active and inactive sites of transcription in real time. In

    comparison with the less intense bursts observed using a similar approach in bacteria

    (Golding et al., 2005), the authors found that the bursts were less frequent but longer lasting.

    In contrast with earlier bacterial models, this shows that bursts in gene expression are the

    primary intrinsic cause of cell-to-cell variability.

    One can also measure mRNA numbers in single cells across a population using variants of

    fluorescence in situ hybridization (FISH) capable of detecting individual mRNA molecules

    (Femino et al., 1998; Raj et al., 2006; Raj et al., 2008). Raj et al. (2006) combined single

    molecule FISH with statistical analysis to show that individual mammalian cells transcribeda stably integrated transgene in infrequent but potent bursts, resulting in large cell-to-cell

    variations in mRNA number (Figures 3C and 3D) that correlated with the presence or

    absence of active sites of transcription [seen also by (Voss et al., 2006)]. These bursts were

    correlated between genes that were located proximally to each other but not between genes

    that were distally located, providing another clue that chromatin remodeling may be

    responsible for genes transitioning between an active and inactive state: opening of the

    chromatin surrounding one gene is likely to open chromatin for neighboring genes, leading

    to correlations in their expression, whereas distant genes are not affected in this coordinated

    manner, resulting in uncorrelated expression. This behavior is also seen in globin expression

    (de Krom et al., 2002) and shows that genomic position can be important in interpreting the

    concepts of intrinsic and extrinsic noise.

    Quantitative single-cell RT-PCR methods have been used to obtain cell-by-cell counts ofendogenous mRNAs, thus circumventing issues associated with generating transgenic cell

    lines and organisms. By simultaneously measuring the numbers of five transcripts in

    individual pancreatic islet cells, Bengtsson et al. (2005) showed that the distributions of

    these mRNAs across the population were heavily skewed as in Figure 3D. Moreover, they

    measured correlations in the fluctuations in the expression of these genes, finding that two

    functionally related genes were highly correlated whereas the rest were uncorrelated,

    perhaps pointing to the existence of common regulators for the two genes. Such findings

    highlight the potential use of stochastic gene expression in uncovering the mechanisms of

    transcriptional regulation. One difficulty with this approach is the rigorous set of controls

    required to calibrate RT-PCR results in molecular units, a problem that can be obviated

    through the use of so-called digital RT-PCR. This method, in which cDNA reverse

    transcribed from an individual cell is fractionated into enough individual PCR reactions that

    each reaction will contain either 0 or 1 cDNAs, has been used to examine the expression ofthe PU.1 transcription factor in both hematopoetic stem cells and in myeloid progenitor cells

    (Warren et al., 2006), in which the authors observed marked heterogeneity in transcript

    levels.

    Atlhough the evidence for transcriptional bursting continues to accumulate, little is known

    about the source of these bursts. As mentioned earlier, one possibility often posited is that

    Raj and van Oudenaarden Page 5

    Cell. Author manuscript; available in PMC 2011 June 18.

    NIH-PAA

    uthorManuscript

    NIH-PAAuthorManuscript

    NIH-PAAuthor

    Manuscript

  • 7/31/2019 Stchstc Gene Expn

    6/21

    stochastic events of chromatin remodeling could underlie the bursts by causing the gene to

    switch between transcriptionally active and inactive states (Becskei et al., 2005; Raj et al.,

    2006; Raser and O'Shea, 2004; Warren et al., 2006). In support of this view, direct

    visualization of chromatin remodeling has shown it to be a slow process that can act over a

    long range on a timescale of hours (Tumbar et al., 1999). However, there are other plausible

    mechanisms that might underlie transcriptional bursts. One possibility is the existence of

    pre-initiation complexes that form on the promoter region of the DNA and facilitate multiple

    rounds of RNA polymerase II transcription events (Blake et al., 2006; Blake et al., 2003). Ifsuch complexes exist only for short periods of time, they could also result in pulsatile

    transcription. Another point to consider is that transcription doesn't take place in a uniform

    fashion throughout the genome but is concentrated in transcriptional factories (Jackson et

    al., 1993; Wansink et al., 1993) to which active genes are recruited (Osborne et al., 2004).

    Remarkably, it appears that a limited number of these factories (on the order of hundreds)

    are responsible for most mRNA transcription in the cell; thus, competition for these factories

    could result in the stochastic expression of any given gene. Ultimately, understanding the

    biochemical origins of bursting may require the application of new (or perhaps combinations

    of old) techniques for imaging gene expression and genome organization in real-time, as

    cell-to-cell variability in population snapshots may not be sufficient to resolve the

    dynamics of the bursting mechanisms (Pedraza and Paulsson, 2008). Although difficult, the

    prize for such a technical feat would be a much deeper understanding of the transcriptional

    process.

    The above studies examining mRNA copy number variation provide insights into the origins

    of noise, although they mostly fail to show how those mRNA fluctuations propagate to noise

    in protein levels. To examine noise in protein levels in human cells, Sigal et al. (2006) used

    a clever strategy to fluorescently tag endogenous proteins. They transfected a cell line with

    DNA containing artificial YFP exons that occasionally insert themselves into an intron, YFP

    is included in the protein encoded by the encapsulating gene. Using time-lapse microscopy,

    the authors were able to show that gene expression in individual cells was variable, but that

    the fluctuations were slowly varying in time; that is, it took multiple cell divisions before a

    highly expressing cell would become a lowly expressing cell and vice versa. Interestingly,

    they also found correlations between genes in the same pathway, but not between unrelated

    genes, echoing the results of Bengtsson et al. (2005).

    Yet, the variability observed at the protein level by Sigal et al. (2006) seems generally much

    smaller than that observed at the mRNA level in the aforementioned studies, with the

    distribution of mRNAs being much more heavily skewed (Figure 3B). How might such a

    discrepancy be resolved? One answer may be methodological: by screening for cells

    expressing a detectable amount of YFP, the proteins with YFP insertions obtained by Sigal

    et al. may be biased towards heavily or constitutively expressing genes with less variability,

    an interpretation supported by the fact that variability in the number of GAPDH mRNAs is

    lower than other genes (Warren et al., 2006). It is also possible that protein stability plays a

    role in the relationship between mRNA and protein variability (Raj et al., 2006). Short-lived

    proteins will track mRNA levels very closely, leading to protein distributions that resemble

    (and correlate strongly with) mRNA distributions. However, if the proteins degrade slowly

    (as is the case for YFP), then the large pool of older proteins will buffer the rapid

    fluctuations in mRNA; that is, mRNA bursts may serve only to top up protein levels. Inthis case, mRNA and protein levels do not strongly correlate.

    Networked noise

    Building on these studies elucidating the sources and characteristics of noise, researchers

    went on to study the effects of noise in simple synthetic genetic networks. One example is

    Raj and van Oudenaarden Page 6

    Cell. Author manuscript; available in PMC 2011 June 18.

    NIH-PAA

    uthorManuscript

    NIH-PAAuthorManuscript

    NIH-PAAuthor

    Manuscript

  • 7/31/2019 Stchstc Gene Expn

    7/21

    transcriptional cascades, which are a common regulatory motif, particularly in development.

    First, researchers investigated the effect of noise in an upstream gene on noise in a

    downstream gene. This was done using multiple fluorescent reporters to quantify the relative

    contributions of variability in the upstream gene, global noise due to effects such as cell size,

    and also noise intrinsic to the expression of the downstream gene (Pedraza and van

    Oudenaarden, 2005; Rosenfeld et al., 2005). They found that variability can be transmitted

    from the upstream gene to the downstream gene, adding substantially to the noise inherent in

    downstream gene's expression. Further study of cascades showed that longer geneticcascades can filter out rapid fluctuations at the expense of amplifying noise in the timing of

    the propagated signal (Hooshangi et al., 2005). Mathematical analysis has also shown that

    stochastic behavior can have the counterintuitive effect of actually lowering transmitted

    variability (Paulsson and Ehrenberg, 2000; Thattai and van Oudenaarden, 2002).

    Negative or positive feedback are other very common types of regulation in genetic

    networks. In these types of feedback loops the protein encoded by a gene negatively or

    positively influences its own transcription. Negative feedback can reduce the effects of noise

    because fluctuations above and below the mean are pushed back towards the mean, as has

    been predicted theoretically (Savageau, 1974; Thattai and van Oudenaarden, 2001) and

    demonstrated experimentally (Austin et al., 2006; Becskei and Serrano, 2000; Dublanche et

    al., 2006).

    In the presence of positive feedback, noise can result in much more dramatic behavior.

    Positive feedback can act as a switch, in which a small amount of expression from a given

    gene can serve to further activate expression of the gene itself, eventually flipping the gene

    from an off state to an on state. In the presence of cooperativity, though, a cell can

    remain in the off state indefinitely, as cooperativity creates a threshold that the protein

    level must surpass in order to trigger the feedback. In that case, occasional large fluctuations

    in gene expression can serve to randomly activate the switch and push the cell into the on

    state (Hasty et al., 2000). This bistable expression pattern has been observed in synthetic

    systems with positive feedback switches (Becskei et al., 2001; Gardner et al., 2000; Isaacs et

    al., 2003; Kramer and Fussenegger, 2005) and also in several naturally occurring genetic

    positive feedback loops (Acar et al., 2005; Maamar and Dubnau, 2005; Maamar et al., 2007;

    Suel et al., 2006; Suel et al., 2007; Smits et al., 2005). The existence of multiple phenotypic

    profiles also appears in more complex biological networks, as we shall see in the nextsection.

    Noise in its natural context

    Researchers have only recently begun to explore the role fluctuations play in biological

    situations. One can imagine two roles for noise in cellular function: one is as a nuisance that

    serves as an impediment to reliable behavior, and one is as a source of variability that cells

    may exploit. In the remainder of the review, we first focus on cases where noise is beneficial

    and then discuss the potential negative effects of noise, drawing on examples in organisms

    ranging from microbes to metazoans.

    Useful unicellular variability

    In unicellular organisms, one can make the argument that variability could be very useful inthat it would allow heterogenous phenotypes even in clonal populations, enabling a

    population of organisms to commit certain subpopulations to different behaviors.

    Variability in a population is enhanced by networks that can produce multiple, mutually

    exclusive profiles of gene expression profiles (such as ON and OFF expression of a

    particular gene) within single organisms. These states are bistable (or multistable) in the

    sense that small variations in expression are insufficient to cause the organism to flip from

    Raj and van Oudenaarden Page 7

    Cell. Author manuscript; available in PMC 2011 June 18.

    NIH-PAA

    uthorManuscript

    NIH-PAAuthorManuscript

    NIH-PAAuthor

    Manuscript

  • 7/31/2019 Stchstc Gene Expn

    8/21

    one state to another and are often heritable, providing a mechanism for epigenetic

    inheritance (Ptashne, 2007). Occasionally, however, a large stochastic fluctuation in gene

    expression can induce a transition from one state to another, an idea that underlies many of

    the following studies.

    MetabolismMetabolic networks are an important and perhaps surprising class of geneticnetworks exhibiting multistability with stochastic transitions. Despite being some of the

    most extensively studied gene networks in existence, only recently have researchers begunto examine the behavior of metabolic genes at the single-cell level, yielding unanticipated

    results. For instance, following up on the pioneering studies of Novick and Weiner, it was

    found that the lactose utilization network inE. coli displayed an all or none type of

    behavior and that single cells stochastically transition between these two states (Mettetal et

    al., 2006; Ozbudak et al., 2004). Such behavior has also been seen in cells that were all

    initially in an uninduced state, arguing that some stochastic mechanism must have caused

    the network to switch from the off to the on expression state. In another example the

    galactose utilization network in yeast also displays strikingly bimodal patterns in the

    expression of the GAL family of genes responsible for galactose metabolism (Acar et al.,

    2005). The authors explained this using a model in which fluctuations in the GAL3 gene

    were responsible for transitions between the ON and OFF states. Then they altered the

    expression of a key feedback component of the network, thereby changing the degree to

    which the fluctuations were buffered and thus modulating the frequency of the stochastictransitions. The dynamics of these switching events has also been analyzed using time-lapse

    microscopy, yielding fascinating results (Kaufmann et al., 2007). There, the authors found

    that not only were the states themselves heritable, but the transition itself was heritable in

    that related yeast cells appeared to switch in a correlated fashion. Again, the authors were

    able to explain their results using a stochastic model, with the key feature being stochastic

    bursts in GAL80 expression. In all of these studies, however, the link between stochastic

    switching and stochastic gene expression has been implicit rather than explicit, with more

    experiments being required to validate the models.

    Of course, these results raise the inevitable question of why genetically identical populations

    would display such marked phenotypic variability in their metabolic pathways. One idea is

    that having individual cells stochastically switch between activating or inactivating a

    metabolic pathway could confer a fitness advantage to the overall population in fluctuatingenvironments (Kussell and Leibler, 2005; Thattai and van Oudenaarden, 2004; Wolf et al.,

    2005). Intuitively, this benefit arises from a tradeoff between anticipation and sensing of

    food sources. Cells can either directly sense food in the environment before activating their

    metabolic networks, or they can choose to stochastically commit some fraction of the

    population to having those metabolic networks active in anticipation of the arrival of a new

    food source. The cost of the former strategy is slow response time and implementation of the

    sensing apparatus, whereas the latter strategy essentially sacrifices some fraction of the

    population to suboptimal growth. These studies have shown that stochastic switching is a

    viable alternative to sensing and that it is most effective when the switching rate is closely

    tuned to the rate at which the environment fluctuates. Experimentally, Acar et al. (2008)

    tested these theories by monitoring the growth rate of a yeast strain with a controllable rate

    of switching in a periodically fluctuating environment and show that fast switchers do

    indeed grow faster in rapidly fluctuating environments whereas slow switchers do better

    when environmental changes come more slowly. Furthermore, Blake et al. (2006) showed

    that expression variability, even in the absence of discrete fit and unfit expression states, can

    be beneficial in times of stress. It is likely, however, that in real biological systems, cells

    rely on some combination of variability in gene expression and sensing in their stress

    responses; elucidating this interplay in biological contexts could have broad implications for

    microbial growth strategies.

    Raj and van Oudenaarden Page 8

    Cell. Author manuscript; available in PMC 2011 June 18.

    NIH-PAA

    uthorManuscript

    NIH-PAAuthorManuscript

    NIH-PAAuthor

    Manuscript

  • 7/31/2019 Stchstc Gene Expn

    9/21

    Microbial stress responsesAnother case of bet-hedging in microbial populations is

    in the response to cellular stress, such as lack of food or exposure to antibiotics. A

    particularly nice example of the former that has garnered considerable recent attention is the

    phenomenon of competence inB. subtilis. B. subtilis has the remarkable ability to take up

    DNA from the environment (called competentence), which exhibits itself upon the entry to

    stationary phase through the activation of a quorum sensing mechanism. Interestingly, only

    a small fraction (roughly 10-20%) of the cells become competent, while the rest remain in a

    vegetative state. This phenotypic variability was first observed over 40 years ago (Nesterand Stocker, 1963) and is the result of a positive feedback loop in which the transcription

    factor ComK promotes its own expression: when the feedback loop is activated, high levels

    of ComK are produced, activating a host of downstream genes involved in DNA uptake,

    whereas non-competent cells produce only low basal amounts of ComK (Maamar and

    Dubnau, 2005; Smits et al., 2005; Suel et al., 2006). The resulting bimodal expression

    pattern is easily visualized using fluorescent proteins.

    A natural hypothesis is that spontaneous fluctuations in comKexpression of sufficient

    magnitude can cause a non-competent cell to transition to competence. To test this notion,

    Maamar et al. (2007) quantified comKexpression in individual non-competent cells by using

    single-molecule FISH to count the number ofcomKtranscripts. They showed that increasing

    the mean level ofcomKtranscription resulted in an increase in the percentage of competent

    cells, presumably because the fraction of cells with fluctuations in ComK above a certainthreshold also increased. To test that possibility directly, they increased the comK

    transcription rate while lowering the translation rate, which reduces noise in gene expression

    while leaving the mean expression level unchanged (Ozbudak et al., 2002; Thattai and van

    Oudenaarden, 2001). Lowering the noise should reduce the number of cells whose

    fluctuations cross the threshold for competence, and indeed the authors found that the

    number of competent cells was dramatically reduced, demonstrating the importance and

    utility of noise theory in biological situations. Another recent study (Suel et al., 2007)

    showed that reducing total cellular noise also resulted in a lower percentage of competent

    cells. To achieve this overall noise reduction, they used a special mutant that is unable to

    septate, resulting in very large cells with multiple genomes. In these large cells, the impact

    of all fluctuations is reduced, given that the cell is in some ways the average of many

    smaller cells, with ever larger cells consequently having ever lower overall fluctuations. The

    authors found that these larger cells did in fact display commensurately fewer transitions tothe competent state. Overall, though, it is important to note that the low number of comK

    transcripts measured (Maamar et al., 2007) and the non-uniformity of the duration of

    competence episodes (Suel et al., 2007) imply that this system has evolved to be

    purposefully imprecise, a feature that cells may exploit in other situations.

    Stochastic effects coupled with positive feedback can also lead to variability in the timing of

    particular molecular events such as the onset of meiosis in yeast (Nachman et al., 2007). In

    this work the timing between introduction of environmental stress and the onset of meiosis

    in individual cells was highly variable. This variability seemed not to depend on position in

    the cell cycle or other external factors, but rather was heavily dependent on noise in the

    expression of the meiotic regulator Ime1 (although cell-size did appear to have a strong

    effect). Together, these studies paint a picture in which noise in gene expression can lead to

    random fates at random times when stressed, a surprising finding that may ultimately proveremarkably prevalent.

    PathogensHeterogeneous phenotypes in clonal populations can also be medically

    relevant. One example is bacterial persistence in the face of antibiotic exposure. Persistent

    cells grow at a much slower rate than non-persistent cells, but are able to survive antibiotic

    treatment. The existence of persistent subpopulations ofMycobacterium tuberculosis,

    Raj and van Oudenaarden Page 9

    Cell. Author manuscript; available in PMC 2011 June 18.

    NIH-PAA

    uthorManuscript

    NIH-PAAuthorManuscript

    NIH-PAAuthor

    Manuscript

  • 7/31/2019 Stchstc Gene Expn

    10/21

    Staphylococcus aureus, and Pseudomonas aeruginosa among other is thought to be a major

    obstacle to effective treatment (Stewart et al., 2003). Interestingly, the work of Balaban et al.

    (2004) showed that a small persistent subpopulation exists even in untreated cultures ofE.

    coli and that these persistent cells are generated continuously during growth. Although not

    much is known about how the underlying network can result in such disparate phenotypes, it

    is entirely possible that stochastic gene expression could play a significant role in

    establishing non-genetic heterogeneity in these populations.

    Another example of heterogeneity in a pathogen is that of the latent phase of HIV infection.

    Upon infection with the HIV virus, a small pool of latent CD4 T lymphocytes forms

    containing stably integrated but non-expressing virus. The low level of expression of the

    virus in this population of cells renders them difficult to target pharmacologically, making

    latency a serious impediment to effective treatment. Weinberger et al. (2005) showed that

    one explanation for the latent and active expression patterns is a positive feedback loop

    mediated by the Tat protein. They showed that stochastic fluctuations in Tat expression can

    interact with the feedback loop to create populations of cells with high and low levels of

    viral expression Interestingly, though, later work (Weinberger et al., 2007) showed that Tat

    positive feedback did not serve to maintain the ON state (as in the competence network in

    B. subtilis) but rather that the heterogeneity was caused by large transient bursts of

    expression that positive feedback served to amplify rather than stabilize (Weinberger et al.,

    2008).

    Random Developments

    As the above examples demonstrate, there is a clear rationale for using stochastic gene

    expression to create a diversity of phenotypes, namely that isogenic populations of viruses,

    bacteria and yeast cannot display heterogeneity in any other way. However, in many higher

    eukaryotes, population diversity largely arises from genetic and environmental diversity,

    making the argument for utilizing stochastic gene expression less plausible. In development,

    for example, one would imagine that a deterministic execution of the developmental

    program would be critical to producing functional tissues, with organism-to-organism

    variations reflecting genetic rather than stochastic differences. Yet even in development,

    researchers are finding many interesting examples of stochastic cell-fate decisions linked to

    stochastic gene expression.

    One celebrated example of stochastic gene expression having an important role in

    development is the expression of different odorant receptors in different sensory neurons in

    mice. Olfaction presents an interesting regulatory challenge, as there are over a thousand

    different odorant receptors, each of which must be expressed differentially in individual

    neurons to confer distinctive sensitivity. Developing a regulatory network capable of such

    complex decision making is prohibitively complex, so the mouse adopts a much simpler

    Monte-Carlo strategy in which each neuron randomly expresses a particular odorant

    receptor (Vassar et al., 1993) in a mutually exclusive fashion (Tsuboi et al., 1999). A

    fascinating line of further inquiry would be to determine the stochastic mechanisms

    responsible for these choices during the development of the olfactory epithelium and

    elucidation of the network responsible for locking in a particular decision once made.

    Another particularly nice instance in which stochastic gene expression has been explicitly

    linked to a cell fate decision is photoreceptor expression in Drosophila eyes. The Drosophila

    eye consists of a large number of optical units called ommatidia, each of which contains two

    cells that in turn express one of a specific pair of photoreceptors, eitherRh3 andRh5 (for

    blue sensitive ommatidia) orRh4 andRh6(for yellow sensitive ommatidia). Wernet et al.

    (2006) showed that this decision is almost exclusively due to the stochastic expression of the

    Raj and van Oudenaarden Page 10

    Cell. Author manuscript; available in PMC 2011 June 18.

    NIH-PAA

    uthorManuscript

    NIH-PAAuthorManuscript

    NIH-PAAuthor

    Manuscript

  • 7/31/2019 Stchstc Gene Expn

    11/21

    spineless gene during mid-pupation, with stochastically large levels ofspineless expression

    resulting in the adoption of the yellow fate in roughly 70% of the ommatidia.

    The process of hematopoiesis, in which progenitor stem cells differentiate into the various

    types of blood cells, is another example in which cellular differentiation may be stochastic

    (Enver et al., 1998; Hume, 2000). To link this stochastic differentiation to variations in gene

    expression, Chang et al. (2008) showed that variability in the expression of the stem cell

    marker Sca-1 in individual cells correlated strongly with the probability of that cell tochoose an erythroid or myeloid lineage. Moreover, microarray analysis on the populations of

    cells expressing high and low levels of Sca-1 showed transcriptome-wide variability,

    indicating that the fluctuations were not limited only to a small set of genes. It would be

    interesting to see how widespread these massively correlated fluctuations are in other

    examples of stochastic differentiation and if these correlations stem from an unknown

    master regulator or arise from noise in many parts of a large interlocking genetic network.

    Shutting out the noise

    Despite these examples of organisms exploiting noise, it is possible, if not probable, that

    noise in gene expression is more generally an obstacle that organisms must overcome to

    achieve robust function. Less is currently known about the mechanisms by which the effects

    of noise are minimized, likely due to the difficulty in studying a phenomenon that by

    definition is invariant to perturbations. In fact, much of the focus on the benefits of noise

    reflects the fact that studying the consequences of stochastic gene expression is much easier

    when the phenomenon in question is itself stochastic. Nevertheless, progress has been made

    in understanding how organisms tolerate noise, from the basics of cellular function through

    development.

    Genomic approaches

    One way to find evidence for the deleterious effects of noise is to make comprehensive

    measurements of noise over a large number of genes and look for evidence that noise has

    been selected against in certain sets of genes. This was the approach taken by Newman et al.

    (2006) and Bar-Even et al. (2006), with the former measuring the noise in expression in over

    2500 genes in yeast and the latter examining fewer genes (43) but in a variety of

    environmental conditions. Both studies reached strikingly similar conclusions, finding thatnoise stemmed mostly from randomness in mRNA synthesis and destruction and that genes

    with higher levels of expression generally exhibited less variability from cell to cell. This

    latter point highlights a potential tradeoff between the level of noise in gene expression and

    the metabolic cost of maintaining a large number of proteins. They also found that stress

    response genes, which are typically non-essential, tended to be noisy, reflecting the potential

    benefits of noise in this class of gene (Blake et al., 2006). In contrast, genes involved in

    protein synthesis and degradation were much less variable, implying that genes essential for

    cellular function require more precise expression levels. The regularity of these essential

    genes may be achieved in a number of ways such as genomic positioning of essential genes

    in areas of open chromatin that are presumably less noisy (Batada and Hurst, 2007). This

    correlative data does not prove the case, however, and an explicit test that noise in essential

    genes is deleterious would be fruitful in this regard.

    Noise minimization and compensation in gene networks

    Given that genes often interact in networks, it is also important to understand how the

    effects of noise are minimized in particular genetic networks. To study this more complex

    problem, Kollman et al.(2005) in their study of the chemotaxis network of E. coli, began

    with several plausible biochemical models of chemotaxis. Each model possessed the

    Raj and van Oudenaarden Page 11

    Cell. Author manuscript; available in PMC 2011 June 18.

    NIH-PAA

    uthorManuscript

    NIH-PAAuthorManuscript

    NIH-PAAuthor

    Manuscript

  • 7/31/2019 Stchstc Gene Expn

    12/21

    fundamental property of precise adaptation of pathway activity to local food signals, but

    varied in their ability to tolerate noise. Upon measuring the noise and correlations in the

    expression of several key components of the pathway, they found that the model that most

    successfully tolerated such noise described a network similar to the one found in E. coli,

    suggesting that the endogenous network may have evolved to tolerate noise while avoiding

    the costs associated with high levels of protein expression.

    Another example of noise-resistance in a signaling pathway is the mating pheromoneresponse pathway in yeast studied by Colman-Lerner et al. (2005). Through the use of dual

    reporters inspired by Elowitz et al. (2002), they quantified all the different sources of cell-to-

    cell variability in their system, with the primary distinction being between random

    biochemical events in the propagation of the signal itself and preexisting differences in cells'

    capacity to respond to the signal. They found that most of the variability observed was due

    to preexisting cellular differences, corroborating other claims that variability in yeast is

    largely extrinsic (Raser and O'Shea, 2004; Volfson et al., 2006). Interestingly, though, they

    found a surprising negative correlation between the signaling capacity of the pathway in

    individual cells and the capacity to express the pathway's target gene in those same cells.

    The implication is that variability in the signaling pathway is compensated for at the level of

    gene expression, thus allowing the cell to produce a robust gene expression profile despite

    large differences in signaling capacities.

    Noise resistance has also driven much research into the networks underlying circadian

    rhythms, biochemical oscillations present in organisms ranging from cyanobacteria to

    humans that are entrained by periodic exposure to sunlight but are capable of free-running

    without any external signals. These oscillations display a remarkable fidelity in their

    duration from cycle to cycle, but the source of this reliability in still unclear and may depend

    on properties of the network used to implement the oscillator. For instance cyanobacteria,

    despite possessing perhaps the simplest known clock, produce very regular oscillations.

    Notably, the proteins involved can oscillate in vitro in the absence of any transcriptional

    regulation at all (Nakajima et al., 2005), but presumably variability in the numbers of these

    proteins in individual cells can cause cells to lose synchrony. Indeed, gene expression

    variability during the clock cycle has many interesting properties (Chabot et al., 2007).

    Another possibility is that cell-cell communication might allow cells to compensate for the

    fluctuations in the oscillations of individual cells. This is not the case in cyanobacteria,however, given that when one places two cells at different phases of the circadian cycle next

    to each other, their progeny robustly maintain the two different cycles inherited from the

    parents (Mihalcescu et al., 2004).

    In higher organisms, transcriptional regulation plays a key role in the generation of circadian

    rhythms, and single cell experiments have shown the performance of the clock in individual

    mammalian cells can be rather poor, with strikingly variable periods observed both in

    culture (Nagoshi et al., 2004) and in whole organisms (Liu et al., 1997). There is some

    evidence behind the general consensus that cell-cell communication allow all the cells in an

    organism's pacemaker to maintain its phase (Liu et al., 1997), but it would be interesting to

    explore how noise in gene expression contributes to dephasing individual cells, especially

    given recent theories claiming that even these networks seem to have some noise-resistant

    properties (Barkai and Leibler, 2000; Forger and Peskin, 2005). More generally, such resultscould apply to other kinds of genetic oscillators like the cell-cycle, where recent work has

    shown that noise is a key factor in cell-cycle timing variability (Di Talia et al., 2007).

    Noise minimization in development

    So far, little work has been done on the role of noise in gene expression in development,

    probably due to difficulties in obtaining quantitative measurements. However, one excellent

    Raj and van Oudenaarden Page 12

    Cell. Author manuscript; available in PMC 2011 June 18.

    NIH-PAA

    uthorManuscript

    NIH-PAAuthorManuscript

    NIH-PAAuthor

    Manuscript

  • 7/31/2019 Stchstc Gene Expn

    13/21

    example of a developmental buffer against noise is the activity of Hsp90 in Arabidopsis

    (Queitsch et al., 2002), the inhibition of which reveals the effects of genetic and

    environmental variability. Surprisingly, this same inhibition results in marked

    developmental variability even in relatively isogenic populations, most likely stemming

    from stochastic effects. An exciting avenue for further research would be to try and link

    stochastic gene expression to phenotypic diversity (familiar to geneticists as the common

    phenomena of partial penetrance or variable expressivity of phenotypes).

    Stochastic gene expression and aging

    Another line of evidence that noise is undesirable comes from research showing that aging is

    correlated with increased noise in gene expression. In one case, researchers showed that the

    expression of a variety of housekeeping and cell-type specific genes in individual murine

    cardiac myocytes become increasingly stochastic as the organism aged (Bahar et al., 2006).

    They further found that treating cells isolated from young animals with hydrogen peroxide

    also produces an increase in expression variability, perhaps indicating that oxidative damage

    may be a factor. Similar stochastic effects have been seen in aging murine muscle tissues

    (Newlands et al., 1998).

    Conversely, the stochastic expression of a gene may actually be responsible for determining

    lifespan in C. elegans (Rea et al., 2005). The authors found that the level of expression of a

    reporter expressed from a heat shock promoter in response to environmental stress on thefirst day of adulthood was remarkably stochastic and moreover predicted the lifespan of the

    organism. Although the mechanisms underlying these stochastic phenomena are still

    unclear, it is possible that aging may be surprisingly dependent on the effects of stochastic

    gene expression.

    Conclusion

    We would like to emphasize that despite the flurry of activity in the area of stochastic gene

    expression over the last several years, the field is still remarkably young, with many

    significant discoveries likely to come in the future. Basic measurements of cell-to-cell

    variability in higher eukaryotes are still in their infancy, and single-molecule techniques

    have shown that surprises still lurk even in supposedly well-characterized systems such asE.

    coli. Moving forward, researchers have also started to examine biological consequences of

    noisealready, there are more and more examples of noise being beneficial in isogenic

    populations, a trend we expect to continue. We anticipate more studies highlighting how

    cells control and tolerate noise to produce reliable behavior. Of course, the most exciting

    discoveries are those that are completely unexpected, and given the fundamental nature of

    stochastic gene expression, it may prove important in unpredictable ways in experimental

    systems both new and old.

    Acknowledgments

    We would like to thank Michael Laub, Ido Golding, Jim Collins and Jeff Gore for many helpful comments on the

    manuscript. We also apologize to any authors whose work we were unable to mention due to space constraints.

    A.v.O was supported by NSF grant PHY-0548484 and NIH grants R01-GM068957 and R01-GM077183. A.R. is

    supported by NSF Fellowship DMS-0603392 and a Burroughs Wellcome Fund Career Award at the ScientificInterface.

    References

    Acar M, Becskei A, van Oudenaarden A. Enhancement of cellular memory by reducing stochastic

    transitions. Nature. 2005; 435:228232. [PubMed: 15889097]

    Raj and van Oudenaarden Page 13

    Cell. Author manuscript; available in PMC 2011 June 18.

    NIH-PAA

    uthorManuscript

    NIH-PAAuthorManuscript

    NIH-PAAuthor

    Manuscript

  • 7/31/2019 Stchstc Gene Expn

    14/21

    Acar M, Mettetal JT, van Oudenaarden A. Stochastic switching as a survival strategy in fluctuating

    environments. Nat Genet. 2008; 40:471475. [PubMed: 18362885]

    Arkin A, Ross J, McAdams HH. Stochastic kinetic analysis of developmental pathway bifurcation in

    phage lambda-infected Escherichia coli cells. Genetics. 1998; 149:16331648. [PubMed: 9691025]

    Austin DW, Allen MS, McCollum JM, Dar RD, Wilgus JR, Sayler GS, Samatova NF, Cox CD,

    Simpson ML. Gene network shaping of inherent noise spectra. Nature. 2006; 439:608611.

    [PubMed: 16452980]

    Bahar R, Hartmann CH, Rodriguez KA, Denny AD, Busuttil RA, Dolle ME, Calder RB, ChisholmGB, Pollock BH, Klein CA, Vijg J. Increased cell-to-cell variation in gene expression in ageing

    mouse heart. Nature. 2006; 441:10111014. [PubMed: 16791200]

    Balaban NQ, Merrin J, Chait R, Kowalik L, Leibler S. Bacterial persistence as a phenotypic switch.

    Science. 2004; 305:16221625. [PubMed: 15308767]

    Bar-Even A, Paulsson J, Maheshri N, Carmi M, O'Shea E, Pilpel Y, Barkai N. Noise in protein

    expression scales with natural protein abundance. Nat Genet. 2006; 38:636643. [PubMed:

    16715097]

    Barkai N, Leibler S. Circadian clocks limited by noise. Nature. 2000; 403:267268. [PubMed:

    10659837]

    Batada NN, Hurst LD. Evolution of chromosome organization driven by selection for reduced gene

    expression noise. Nat Genet. 2007; 39:945949. [PubMed: 17660811]

    Beach DL, Salmon ED, Bloom K. Localization and anchoring of mRNA in budding yeast. Curr Biol.

    1999; 9:569578. [PubMed: 10359695]Becskei A, Kaufmann BB, van Oudenaarden A. Contributions of low molecule number and

    chromosomal positioning to stochastic gene expression. Nat Genet. 2005; 37:937944. [PubMed:

    16086016]

    Becskei A, Seraphin B, Serrano L. Positive feedback in eukaryotic gene networks: cell differentiation

    by graded to binary response conversion. Embo J. 2001; 20:25282535. [PubMed: 11350942]

    Becskei A, Serrano L. Engineering stability in gene networks by autoregulation. Nature. 2000;

    405:590593. [PubMed: 10850721]

    Bengtsson M, Stahlberg A, Rorsman P, Kubista M. Gene expression profiling in single cells from the

    pancreatic islets of Langerhans reveals lognormal distribution of mRNA levels. Genome Res.

    2005; 15:13881392. [PubMed: 16204192]

    Bertrand E, Chartrand P, Schaefer M, Shenoy SM, Singer RH, Long RM. Localization of ASH1

    mRNA particles in living yeast. Mol Cell. 1998; 2:437445. [PubMed: 9809065]

    Blake WJ, Balazsi G, Kohanski MA, Isaacs FJ, Murphy KF, Kuang Y, Cantor CR, Walt DR, CollinsJJ. Phenotypic consequences of promoter-mediated transcriptional noise. Mol Cell. 2006; 24:853

    865. [PubMed: 17189188]

    Blake WJ, M KA, Cantor CR, Collins JJ. Noise in eukaryotic gene expression. Nature. 2003; 422:633

    637. [PubMed: 12687005]

    Cai L, Friedman N, Xie XS. Stochastic protein expression in individual cells at the single molecule

    level. Nature. 2006; 440:358362. [PubMed: 16541077]

    Chabot JR, Pedraza JM, Luitel P, van Oudenaarden A. Stochastic gene expression out-of-steady-state

    in the cyanobacterial circadian clock. Nature. 2007; 450:12491252. [PubMed: 18097413]

    Chang HH, Hemberg M, Barahona M, Ingber DE, Huang S. Transcriptome-wide noise controls

    lineage choice in mammalian progenitor cells. Nature. 2008; 453:544547. [PubMed: 18497826]

    Chubb JR, Trcek T, Shenoy SM, Singer RH. Transcriptional pulsing of a developmental gene. Curr

    Biol. 2006; 16:10181025. [PubMed: 16713960]

    Colman-Lerner A, Gordon A, Serra E, Chin T, Resnekov O, Endy D, Pesce CG, Brent R. Regulatedcell-to-cell variation in a cell-fate decision system. Nature. 2005; 437:699706. [PubMed:

    16170311]

    de Krom M, van de Corput M, von Lindern M, Grosveld F, Strouboulis J. Stochastic patterns in globin

    gene expression are established prior to transcriptional activation and are clonally inherited. Mol

    Cell. 2002; 9:13191326. [PubMed: 12086628]

    Raj and van Oudenaarden Page 14

    Cell. Author manuscript; available in PMC 2011 June 18.

    NIH-PAA

    uthorManuscript

    NIH-PAAuthorManuscript

    NIH-PAAuthor

    Manuscript

  • 7/31/2019 Stchstc Gene Expn

    15/21

    Di Talia S, Skotheim JM, Bean JM, Siggia ED, Cross FR. The effects of molecular noise and size

    control on variability in the budding yeast cell cycle. Nature. 2007; 448:947951. [PubMed:

    17713537]

    Dublanche Y, Michalodimitrakis K, Kummerer N, Foglierini M, Serrano L. Noise in transcription

    negative feedback loops: simulation and experimental analysis. Mol Syst Biol. 2006; 2:41.

    [PubMed: 16883354]

    Elowitz MB, Leibler S. A synthetic oscillatory network of transcriptional regulators. Nature. 2000;

    403:335338. [PubMed: 10659856]

    Elowitz MB, Levine AJ, Siggia ED, Swain PS. Stochastic gene expression in a single cell. Science.

    2002; 297:11831186. [PubMed: 12183631]

    Enver T, Heyworth CM, Dexter TM. Do stem cells play dice? Blood. 1998; 92:348351. discussion

    352. [PubMed: 9657728]

    Femino AM, Fay FS, Fogarty K, Singer RH. Visualization of single RNA transcripts in situ. Science.

    1998; 280:585590. [PubMed: 9554849]

    Forger DB, Peskin CS. Stochastic simulation of the mammalian circadian clock. Proc Natl Acad Sci U

    S A. 2005; 102:321324. [PubMed: 15626756]

    Friedman N, Cai L, Xie XS. Linking stochastic dynamics to population distribution: an analytical

    framework of gene expression. Phys Rev Lett. 2006; 97:168302. [PubMed: 17155441]

    Gardner TS, Cantor CR, Collins JJ. Construction of a genetic toggle switch in Escherichia coli. Nature.

    2000; 403:339342. [PubMed: 10659857]

    Gillespie DT. Exact Stochastic Simulation of Coupled Chemical-Reactions. Journal of PhysicalChemistry. 1977; 81:23402361.

    Golding I, Paulsson J, Zawilski SM, Cox EC. Real-time kinetics of gene activity in individual bacteria.

    Cell. 2005; 123:10251036. [PubMed: 16360033]

    Hasty J, Pradines J, Dolnik M, Collins JJ. Noise-based switches and amplifiers for gene expression.

    Proc Natl Acad Sci U S A. 2000; 97:20752080. [PubMed: 10681449]

    Hooshangi S, Thiberge S, Weiss R. Ultrasensitivity and noise propagation in a synthetic transcriptional

    cascade. Proc Natl Acad Sci U S A. 2005; 102:35813586. [PubMed: 15738412]

    Hume DA. Probability in transcriptional regulation and its implications for leukocyte differentiation

    and inducible gene expression. Blood. 2000; 96:23232328. [PubMed: 11001878]

    Isaacs FJ, Hasty J, Cantor CR, Collins JJ. Prediction and measurement of an autoregulatory genetic

    module. Proc Natl Acad Sci U S A. 2003; 100:77147719. [PubMed: 12808135]

    Jackson DA, Hassan AB, Errington RJ, Cook PR. Visualization of focal sites of transcription within

    human nuclei. Embo J. 1993; 12:10591065. [PubMed: 8458323]Karmakar R, Bose I. Graded and binary responses in stochastic gene expression. Phys Biol. 2004;

    1:197204. [PubMed: 16204839]

    Kaufmann BB, Yang Q, Mettetal JT, van Oudenaarden A. Heritable stochastic switching revealed by

    single-cell genealogy. PLoS Biol. 2007; 5:e239. [PubMed: 17803359]

    Kepler TB, Elston TC. Stochasticity in transcriptional regulation: origins, consequences, and

    mathematical representations. Biophys J. 2001; 81:31163136. [PubMed: 11720979]

    Ko MS. A stochastic model for gene induction. J Theor Biol. 1991; 153:181194. [PubMed: 1787735]

    Ko MS, Nakauchi H, Takahashi N. The dose dependence of glucocorticoid-inducible gene expression

    results from changes in the number of transcriptionally active templates. Embo J. 1990; 9:2835

    2842. [PubMed: 2167833]

    Kollmann M, Lovdok L, Bartholome K, Timmer J, Sourjik V. Design principles of a bacterial

    signalling network. Nature. 2005; 438:504507. [PubMed: 16306993]

    Kramer BP, Fussenegger M. Hysteresis in a synthetic mammalian gene network. Proc Natl Acad Sci U

    S A. 2005; 102:95179522. [PubMed: 15972812]

    Kussell E, Leibler S. Phenotypic diversity, population growth, and information in fluctuating

    environments. Science. 2005; 309:20752078. [PubMed: 16123265]

    Liu C, Weaver DR, Strogatz SH, Reppert SM. Cellular construction of a circadian clock: period

    determination in the suprachiasmatic nuclei. Cell. 1997; 91:855860. [PubMed: 9413994]

    Raj and van Oudenaarden Page 15

    Cell. Author manuscript; available in PMC 2011 June 18.

    NIH-PAA

    uthorManuscript

    NIH-PAAuthorManuscript

    NIH-PAAuthor

    Manuscript

  • 7/31/2019 Stchstc Gene Expn

    16/21

    Maamar H, Dubnau D. Bistability in the Bacillus subtilis K-state (competence) system requires a

    positive feedback loop. Mol Microbiol. 2005; 56:615624. [PubMed: 15819619]

    Maamar H, Raj A, Dubnau D. Noise in gene expression determines cell fate in Bacillus subtilis.

    Science. 2007; 317:526529. [PubMed: 17569828]

    McAdams HH, Arkin A. Stochastic mechanisms in gene expression. Proc Natl Acad Sci U S A. 1997;

    94:814819. [PubMed: 9023339]

    Mettetal JT, Muzzey D, Pedraza JM, Ozbudak EM, van Oudenaarden A. Predicting stochastic gene

    expression dynamics in single cells. Proc Natl Acad Sci U S A. 2006; 103:73047309. [PubMed:16648266]

    Mihalcescu I, Hsing W, Leibler S. Resilient circadian oscillator revealed in individual cyanobacteria.

    Nature. 2004; 430:8185. [PubMed: 15229601]

    Murphy KF, Balazsi G, Collins JJ. Combinatorial promoter design for engineering noisy gene

    expression. Proc Natl Acad Sci U S A. 2007; 104:1272612731. [PubMed: 17652177]

    Nachman I, Regev A, Ramanathan S. Dissecting timing variability in yeast meiosis. Cell. 2007;

    131:544556. [PubMed: 17981121]

    Nagoshi E, Saini C, Bauer C, Laroche T, Naef F, Schibler U. Circadian gene expression in individual

    fibroblasts: cell-autonomous and self-sustained oscillators pass time to daughter cells. Cell. 2004;

    119:693705. [PubMed: 15550250]

    Nakajima M, Imai K, Ito H, Nishiwaki T, Murayama Y, Iwasaki H, Oyama T, Kondo T.

    Reconstitution of circadian oscillation of cyanobacterial KaiC phosphorylation in vitro. Science.

    2005; 308:414415. [PubMed: 15831759]Nester EW, Stocker BA. Biosynthetic Latency In Early Stages Of Deoxyribonucleic

    Acidtransformation In Bacillus Subtilis. J Bacteriol. 1963; 86:785796. [PubMed: 14066476]

    Newlands S, Levitt LK, Robinson CS, Karpf AB, Hodgson VR, Wade RP, Hardeman EC.

    Transcription occurs in pulses in muscle fibers. Genes Dev. 1998; 12:27482758. [PubMed:

    9732272]

    Newman JR, Ghaemmaghami S, Ihmels J, Breslow DK, Noble M, DeRisi JL, Weissman JS. Single-

    cell proteomic analysis of S. cerevisiae reveals the architecture of biological noise. Nature. 2006;

    441:840846. [PubMed: 16699522]

    Novick A, Weiner M. Enzyme Induction as an All-or-None Phenomenon. Proc Natl Acad Sci U S A.

    1957; 43:553566. [PubMed: 16590055]

    Osborne CS, Chakalova L, Brown KE, Carter D, Horton A, Debrand E, Goyenechea B, Mitchell JA,

    Lopes S, Reik W, Fraser P. Active genes dynamically colocalize to shared sites of ongoing

    transcription. Nat Genet. 2004; 36:10651071. [PubMed: 15361872]Ozbudak EM, Thattai M, Kurtser I, Grossman AD, van Oudenaarden A. Regulation of noise in the

    expression of a single gene. Nat Genet. 2002; 31:6973. [PubMed: 11967532]

    Ozbudak EM, Thattai M, Lim HN, Shraiman BI, Van Oudenaarden A. Multistability in the lactose

    utilization network of Escherichia coli. Nature. 2004; 427:737740. [PubMed: 14973486]

    Paulsson J, Ehrenberg M. Random signal fluctuations can reduce random fluctuations in regulated

    components of chemical regulatory networks. Phys Rev Lett. 2000; 84:54475450. [PubMed:

    10990965]

    Paulsson J. Summing up the noise in gene networks. Nature. 2004; 427:415418. [PubMed:

    14749823]

    Pedraza JM, Paulsson J. Effects of molecular memory and bursting on fluctuations in gene expression.

    Science. 2008; 319:339343. [PubMed: 18202292]

    Pedraza JM, van Oudenaarden A. Noise propagation in gene networks. Science. 2005; 307:19651969.

    [PubMed: 15790857]Ptashne M. On the use of the word epigenetic. Curr Biol. 2007; 17:R233236. [PubMed: 17407749]

    Queitsch C, Sangster TA, Lindquist S. Hsp90 as a capacitor of phenotypic variation. Nature. 2002;

    417:618624. [PubMed: 12050657]

    Raj A, Peskin CS, Tranchina D, Vargas DY, Tyagi S. Stochastic mRNA synthesis in mammalian cells.

    PLoS Biol. 2006; 4:e309. [PubMed: 17048983]

    Raj and van Oudenaarden Page 16

    Cell. Author manuscript; available in PMC 2011 June 18.

    NIH-PAA

    uthorManuscript

    NIH-PAAuthorManuscript

    NIH-PAAuthor

    Manuscript

  • 7/31/2019 Stchstc Gene Expn

    17/21

    Raj A, van den Bogaard P, Rifkin SA, van Oudenaarden A, Tyagi S. Imaging individual mRNA

    molecules using sets of singly labeled probes. Nat Meth. September 21.2008 published online. doi

    ##################.

    Raser JM, O'Shea EK. Control of stochasticity in eukaryotic gene expression. Science. 2004;

    304:18111814. [PubMed: 15166317]

    Rea SL, Wu D, Cypser JR, Vaupel JW, Johnson TE. A stress-sensitive reporter predicts longevity in

    isogenic populations of Caenorhabditis elegans. Nat Genet. 2005; 37:894898. [PubMed:

    16041374]

    Ressler KJ, Sullivan SL, Buck LB. A zonal organization of odorant receptor gene expression in the

    olfactory epithelium. Cell. 1993; 73:597609. [PubMed: 7683976]

    Rosenfeld N, Young JW, Alon U, Swain PS, Elowitz MB. Gene regulation at the single-cell level.

    Science. 2005; 307:19621965. [PubMed: 15790856]

    Ross IL, Browne CM, Hume DA. Transcription of individual genes in eukaryotic cells occurs

    randomly and infrequently. Immunol Cell Biol. 1994; 72:177185. [PubMed: 8200693]

    Savageau MA. Comparison of classical and autogenous systems of regulation in inducible operons.

    Nature. 1974; 252:546549. [PubMed: 4431516]

    Shahrezaei V, Ollivier JF, Swain PS. Colored extrinsic fluctuations and stochastic gene expression.

    Mol Syst Biol. 2008; 4:196. [PubMed: 18463620]

    Sigal A, Milo R, Cohen A, Geva-Zatorsky N, Klein Y, Liron Y, Rosenfeld N, Danon T, Perzov N,

    Alon U. Variability and memory of protein levels in human cells. Nature. 2006; 444:643646.

    [PubMed: 17122776]Smits WK, Eschevins CC, Susanna KA, Bron S, Kuipers OP, Hamoen LW. Stripping Bacillus: ComK

    auto-stimulation is responsible for the bistable response in competence development. Mol

    Microbiol. 2005; 56:604614. [PubMed: 15819618]

    Spudich JL, Koshland DE Jr. Non-genetic individuality: chance in the single cell. Nature. 1976;

    262:467471. [PubMed: 958399]

    Stewart GR, Robertson BD, Young DB. Tuberculosis: a problem with persistence. Nat Rev Microbiol.

    2003; 1:97105. [PubMed: 15035039]

    Suel GM, Garvia-Ojalvo J, Liberman LM, Elowitz MB. An excitable gene regulatory circuit induces

    transient cellular differentiation. Nature. 2006; 440:545550. [PubMed: 16554821]

    Suel GM, Kulkarni RP, Dworkin J, Garcia-Ojalvo J, Elowitz MB. Tunability and noise dependence in

    differentiation dynamics. Science. 2007; 315:17161719. [PubMed: 17379809]

    Swain PS, Elowitz MB, Siggia ED. Intrinsic and extrinsic contributions to stochasticity in gene

    expression. Proc Natl Acad Sci U S A. 2002; 99:1279512800. [PubMed: 12237400]Takasuka N, White MR, Wood CD, Robertson WR, Davis JR. Dynamic changes in prolactin promoter

    activation in individual living lactotrophic cells. Endocrinology. 1998; 139:13611368. [PubMed:

    9492073]

    Thattai M, van Oudenaarden A. Intrinsic noise in gene regulatory networks. Proc Natl Acad Sci U S A.

    2001; 98:86148619. [PubMed: 11438714]

    Thattai M, van Oudenaarden A. Attenuation of noise in ultrasensitive signaling cascades. Biophys J.

    2002; 82:29432950. [PubMed: 12023217]

    Thattai M, van Oudenaarden A. Stochastic gene expression in fluctuating environments. Genetics.

    2004; 167:523530. [PubMed: 15166174]

    Tsuboi A, Yoshihara S, Yamazaki N, Kasai H, Asai-Tsuboi H, Komatsu M, Serizawa S, Ishii T,

    Matsuda Y, Nagawa F, Sakano H. Olfactory neurons expressing closely linked and homologous

    odorant receptor genes tend to project their axons to neighboring glomeruli on the olfactory bulb. J

    Neurosci. 1999; 19:84098418. [PubMed: 10493742]Tumbar T, Sudlow G, Belmont AS. Large-scale chromatin unfolding and remodeling induced by VP16

    acidic activation domain. J Cell Biol. 1999; 145:13411354. [PubMed: 10385516]

    Vassar R, Ngai J, Axel R. Spatial segregation of odorant receptor expression in the mammalian

    olfactory epithelium. Cell. 1993; 74:309318. [PubMed: 8343958]

    Volfson D, Marciniak J, Blake WJ, Ostroff N, Tsimring LS, Hasty J. Origins of extrinsic variability in

    eukaryotic gene expression. Nature. 2006; 439:861864. [PubMed: 16372021]

    Raj and van Oudenaarden Page 17

    Cell. Author manuscript; available in PMC 2011 June 18.

    NIH-PAA

    uthorManuscript

    NIH-PAAuthorManuscript

    NIH-PAAuthor

    Manuscript

  • 7/31/2019 Stchstc Gene Expn

    18/21

    Voss TC, John S, Hager GL. Single-cell analysis of glucocorticoid receptor action reveals that

    stochastic post-chromatin association mechanisms regulate ligand-specific transcription. Mol

    Endocrinol. 2006; 20:26412655. [PubMed: 16873444]

    Wansink DG, Schul W, van der Kraan I, van Steensel B, van Driel R, de Jong L. Fluorescent labeling

    of nascent RNA reveals transcription by RNA polymerase II in domains scattered throughout the

    nucleus. J Cell Biol. 1993; 122:283293. [PubMed: 8320255]

    Warren L, Bryder D, Weissman IL, Quake SR. Transcription factor profiling in individual

    hematopoietic progenitors by digital RT-PCR. Proc Natl Acad Sci U S A. 2006; 103:17807

    17812. [PubMed: 17098862]

    Weinberger LS, Burnett JC, Toettcher JE, Arkin AP, Schaffer DV. Stochastic gene expression in a

    lentiviral positive-feedback loop: HIV-1 Tat fluctuations drive phenotypic diversity. Cell. 2005;

    122:169182. [PubMed: 16051143]

    Weinberger LS, Schenk T. An HIV feedback resistor: auto-regulatory circuit deactivator and noise

    buffer. PLoS Biology. 2007; 5(1):e9. [PubMed: 17194214]

    Weinberger LS, Dar RD, Simpson ML. Transient-mediated fate determination in a transcriptional

    circuit of HIV. Nat Genet. 2008; 40:466470. [PubMed: 18344999]

    Wernet MF, Mazzoni EO, Celik A, Duncan DM, Duncan I, Desplan C. Stochastic spineless expression

    creates the retinal mosaic for colour vision. Nature. 2006; 440:174180. [PubMed: 16525464]

    White MR, Masuko M, Amet L, Elliott G, Braddock M, Kingsman AJ, Kingsman SM. Real-time

    analysis of the transcriptional regulation of HIV and hCMV promoters in single mammalian

    cells. J Cell Sci. 1995; 108(Pt 2):441455. [PubMed: 7768992]

    Wolf DM, Vazirani VV, Arkin AP. Diversity in times of adversity: probabilistic strategies in microbial

    survival games. J Theor Biol. 2005; 234:227253. [PubMed: 15757681]

    Xu EY, Zawadzki KA, Broach JR. Single-cell observations reveal intermediate transcriptional

    silencing states. Mol Cell. 2006; 23:219229. [PubMed: 16857588]

    Yarchuk O, Jacques N, Guillerez J, Dreyfus M. Interdependence of translation, transcription and

    mRNA degradation in the lacZ gene. J Mol Biol. 1998; 226:581596. [PubMed: 1507217]

    Yu J, Xiao J, Ren X, Lao K, Xie XS. Probing gene expression in live cells, one protein molecule at a

    time. Science. 2006; 311:16001603. [PubMed: 16543458]

    Raj and van Oudenaarden Page 18

    Cell. Author manuscript; available in PMC 2011 June 18.

    NIH-PAA

    uthorManuscript

    NIH-PAAuthorManuscript

    NIH-PAAuthor

    Manuscript

  • 7/31/2019 Stchstc Gene Expn

    19/21

    Figure 1. Intrinsic and extrinsic contributions to noise in gene expressionA) A fluorescence image of individualE. coli displaying marked cell-to-cell variability in

    the expression of two identically regulated fluorescent proteins. B) Schematic depiction of

    the temporal behaviors of extrinsic noise (upper) and intrinsic noise (lower). C) Expected

    cell-to-cell variations when fluctuations are intrinsic, extrinsic or both. (A and B adapted

    from Elowitz et al., 2002).

    Raj and van Oudenaarden Page 19

    Cell. Author manuscript; available in PMC 2011 June 18.

    NIH-PAA

    uthorManuscript

    NIH-PAAuthorManuscript

    NIH-PAAuthor

    Manuscript

  • 7/31/2019 Stchstc Gene Expn

    20/21

    Figure 2. Noise in prokaryotic gene expression depends on the rates of transcription andtranslation

    A) When the transcription rate is high, variability in protein levels is low, but B) when the

    transcription rate is lowered and the translation rate is raised, gene expression is far noisier,

    even at the same mean, as shown in Ozbudak et al. (2002).

    Raj and van Oudenaarden Page 20

    Cell. Author manuscript; available in PMC 2011 June 18.

    NIH-PAA

    uthorManuscript

    NIH-PAAuthorManuscript

    NIH-PAAuthor

    Manuscript

  • 7/31/2019 Stchstc Gene Expn

    21/21

    Figure 3. The Contribution of Transcriptional Bursts to Cell-to-Cell Variability

    A) Transcription without bursts with a relatively small amount of noise. B) Bursts in

    transcription can cause significantly higher variability, even when producing the same mean

    number of transcripts. C) In situ detection of individual mRNA molecules reveals large cell-

    to-cell variability in mammalian cells. D) Experimental histogram of mRNA numbers. The

    grey dashed line depicts the theoretical distribution one would expect in the absence of

    transcriptional bursts. (C and D adapted from Raj et al., 2006)

    Raj and van Oudenaarden Page 21

    Cell. Author manuscript; available in PMC 2011 June 18.

    NIH-PAA

    uthorManuscript

    NIH-PAAuthorManuscript

    NIH-PAAuthor

    Manuscript